Herbal medicine Gan‑fu‑kang downregulates Wnt/Ca2+ signaling to attenuate liver fibrogenesis in vitro and in vivo

  • Authors:
    • Yujie Jia
    • Lijun Yuan
    • Tingting Xu
    • Hanshu Li
    • Guang Yang
    • Miaona Jiang
    • Caihua Zhang
    • Cong Li
  • View Affiliations

  • Published online on: April 15, 2016     https://doi.org/10.3892/mmr.2016.5148
  • Pages: 4705-4714
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study was designed to verify the effect of the Chinese prescription Gan‑fu‑kang (GFK) on the treatment of liver fibrosis, and to investigate its underlying mechanisms. Liver fibrosis was established in rats by the subcutaneous administration of 0.5 mg/kg carbon tetrachloride (CCl4) twice a week for 8 weeks. Subsequently, the rats were divided into four CCl4 groups, which were treated daily with vehicle and GFK (31.25, 312.5 and 3,125 mg/kg/day) orally between weeks 9 and 20. The inhibitory action of GFK‑medicated serum on platelet‑derived growth factor (PDGF)‑stimulated HSC‑T6 cells was also investigated. Biochemical parameters, hydroxyproline (Hyp) content and histological changes to the liver were measured. Reverse transcription‑quantitative polymerase chain reaction, western blotting and immunohistochemistry were used to examine the expression of α‑smooth muscle actin (α‑SMA), PDGF‑BB, PDGF receptor β, collagen type I and II, and the Wnt/Ca2+ signaling pathway. The results showed that GFK significantly alleviated the histological changes, decreased the content of Hyp in the liver and improved liver function in rats. In addition, GFK and GFK‑medicated serum effectively inhibited collagen deposition, reduced the expression of α‑SMA and downregulated the Wnt/Ca2+ signaling pathway in vivo and in vitro, respectively, as well as cell viability (P<0.05). These results indicated that GFK was effective in attenuating liver injury and fibrosis through downregulation of the Wnt/Ca2+ signaling pathway.

Introduction

Liver fibrosis is a common wound-healing response of the liver to a variety of chronic injuries, including infections, particularly hepatitis B or C, alcoholic steatohepatitis, non-alcoholic steatohepatitis and toxic agents, and is characterized by an excessive deposition of extracellular matrix (ECM) (1,2). It has been demonstrated that activated hepatic stellate cells (HSCs) are the predominant cell type responsible for ECM accumulation in the liver, and their activation is associated with specific cytoskeletal and phenotypic profiles (3,4). Therefore, the majority of antifibrotic therapies are designed to inhibit the activation and proliferation of HSCs. Platelet-derived growth factor (PDGF) is the most potent mitogenic and proliferative cytokine described for HSCs (5). PDGF-BB is the most potent PDGF isoform and has been shown to be the most effective stimulator of HSC proliferation (6). PDGF receptor β (PDGFRβ) amplifies biological responses to PDGF-BB, leading to the activation of downstream signaling pathways in HSCs (7).

The Wnt signaling pathway is essential for embryogenesis and adult tissue maintenance, and disturbance in this signaling promotes neurodegenerative diseases and cancer (810). There are two predominant Wnt signaling pathways: The canonical Wnt/β-catenin pathway and the non-canonical Wnt/Ca2+ pathway. In previous decades, the canonical and non-canonical Wnt signaling pathway have been reported to be important in liver development and remodeling, and HSC activation (1113). Previously, it was reported that the Wnt/Ca2+ signaling pathway can be activated in human HSCs induced by PDGF-BB, and is involved in HSC activation and proliferation (14). However, the role of the non-canonical Wnt signaling pathway in this specific pathophysiological process has received little attention, and remains to be fully elucidated, particularly concerning the Wnt/Ca2+ signaling pathway (15).

Traditional Chinese herbs have been widely used as hepatoprotective and antifibrotic drugs in humans (16) and animal models (17), with novel characteristics, including being multi-ingredient, multi-targeting and with low adverse effects. Gan-fu-kang (GFK) is a complex prescription Chinese herbal medicine composed of 11 medical herbs, including Salvia miltiorrhiza, Astragalus membranaceus, red peony root, white peony root, Radix paeoniae alba and Chinese thorowax root (18). This herbal formula is considered to have effects in increasing blood volume, energy and blood flow to the liver (18). Our pilot study showed that GFK has markedly protective and therapeutic effects in an animal model of hepatic injury induced by carbon tetrachloride (CCl4) (18). Mechanistic investigations have shown that GFK downregulates the mitogen-activated protein kinase/activator protein 1 pathway (19) and canonical Wnt/β-catenin signaling pathway (20) in the fibrotic liver. However, the mechanism by which GFK inhibits liver fibrosis remains to be fully elucidate.

The aim of the present study was to examine the protective effect of GFK on hepatic fibrosis in liver tissues from Sprague-Dawley rats and HSC-T6 cells. Furthermore, the present study aimed to confirm whether GFK attenuates hepatic fibrosis via the Wnt/Ca2+ pathway to elucidate the possible underlying mechanism of its anti-fibrotic effect.

Materials and methods

Herbal medicine composition

GFK consists of 11 herbs, including 30 g each of Salviae miltiorrhizae radix and Milkvetch root; 20 g each of Fructus aurantii and Hoelen; 15 g each of Radix paeoniae rubra, Radix paeoniae alba, Radix angelicae sinensis, Radix rehmanniae and Rhizoma atractylodis macrocephalae; and 10 g each of Radix bupleuri and Radix glycyrrhizae (Table I). The 11 crude drugs were purchased from the pharmacy of the Second Affiliated Hospital of Dalian Medical University (Dalian, China) and extracted by the Department of Pathophysiology of Dalian Medical University. The herbal decoction was stored at −20°C.

Table I

Components of the herbal prescription Gan-fu-kang.

Table I

Components of the herbal prescription Gan-fu-kang.

Herb nameScientific nameLocal namePlace of origin
(China)
Relative
quantity (g)
Salviae miltiorrhizae radixSalvia miltiorrhiza BungeDan ShenHebei30
Milkvetch rootAstragalus membranaceus (Fisch.) BungeHuang QiShanxi30
Fructus aurantiiCitrus aurantium L.ZhiKeHubei20
HoelenPoria cocos (Schw.) WolfFu LingHubei20
Radix paeoniae rubraPaeonia veitchii LynchChi ShaoSichuan15
Radix paeoniae albaPaeonia emodi subsp. sterniana (H.R. Fletcher) HaldaBai ShaoAnhui15
Radix angelicae sinensisAngelica sinensis (Oliv.) DielsDang GuiGansu15
Radix rehmanniaeRehmannia chingii H.L. LiDi HuangLiaoning15
Rhizoma atractylodis macrocephalaeAtractylodes macrocephala Koidz.Bai ZhuZhejiang15
Radix bupleuriBupleurum chinense DC.Chai HuHubei10
Radix glycyrrhizaeGlycyrrhiza uralensis Fisch.Gan CaoNeimenggu10
Animals

Sprague-Dawley rats (n=53; 26 male and 27 female; weight, 180–220 g; age, 6 weeks) were supplied by the Experimental Animal Center of Dalian Medical University [confirmation no. SCXK (Liao) 2004–0017]. The rats were housed in an air-conditioned room at 22±1°C, with 50–60% relative humidity and a 12 h light-dark cycle, and were fed a standard laboratory diet and tap water ad libitum. The experiments were performed in accordance with the principles and guidelines for the human treatment of animals set by the National Institutes of Health Guide (NIH publication no. 85–23, revised 1985) (21) and was approved by the ethics committee of Dalian Medical University.

Experimental model and drug treatment

The rats were randomly divided into five groups: Normal control group (n=12), CCl4 (Tianjin Guangfu Fine Chemical Research Institute, Tianjin, China) model group (n=8) and three GFK groups (n=11). These rats were treated by subcutaneous injection of CCl4 (0.5 mg/kg in a vehicle of olive oil; twice/week) for 8 weeks, with the exception of the rats in the normal control group, which were treated with vehicle only. In the treatment groups, the rats received GFK via oral administration (31.25, 312.5 and 3,125 mg/kg/day) between weeks 9 and 20. The model group and the normal control group were administered with an equal volume of normal saline. All rats were sacrificed by cervical dislocation under diethyl ether (Nanjing Chemical Reagent Co., Ltd., Nanjing, China) anesthesia at the end of week 20. Blood (~4 ml) and complete liver samples were obtained for further examination.

Preparation of drug-medicated serum

A total of 20 rats were randomly divided into two groups. Sera was obtained from the rats, following administration with normal saline or GFK at the middle dose (312.5 mg/kg/day) by gavage for 7 days.

Biochemical determination

The levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (ALB) and globulin in the serum samples were determined using a commercial test reagent (Nanjing Jiancheng Bioengineering Institute, Nanjing, China). ALB was determined in 0.02 ml serum using bromocresol green colorimetry and total protein (TP) was determined by Coomassie brilliant blue colorimetry in 0.05 ml serum. The samples were incubated with either reagent for 10 min at room temperature prior to measuring optical density at 628 nm (bromocresol green) or 595 nm (Coomassie brilliant blue) on a spectrophotometer (721; Shanghai Optical Instrument Factory, Shanghai, China).

Histopathological examination

The rat liver tissues were fixed in 10% formalin (Nanjing Chemical Reagent Co., Ltd.), embedded in paraffin (Sigma-Aldrich, St. Louis, MO, USA), cut (4 μm) and stained with either hematoxylin and eosin (H&E; Nanjing Jiancheng Bioengineering Institute) or with Picric Sirius red (Nanjing Jiancheng Bioengineering Institute). The sections were examined by light microscope (Eclipse 50i; Nikon Corporation, Tokyo, Japan), and the collagen type I and type III staining by Picric Sirius red were observed by polarizing microscope (CX31P; Olympus Corporation, Tokyo, Japan). The liver fibrosis was evaluated by a pathologist in a blinded-manner, and scored and graded according to the method of Scheuer (22), as follows: Stage 0, no fibrosis; stage 1, fibrosis expansion of certain portal areas; stage 2, formation of fibrous septa around the portal area; stage 3, fibrosis with architectural distortion, complete septa interconnecting with each; stage 4, early cirrhosis or cirrhosis.

Hepatic hydroxyproline (Hyp) assay

The content of Hyp was measured spectrophotometrically using a commercially available kit (Nanjing Jiancheng Bioengineering Institute). The quantities of Hyp in the rat liver are expressed as μg/g wet tissue.

Cell culture and 3–4,5-Dimethylthiazol-2-yl)-2,5-dipheny ltetrazolium bromide (MTT) assay

The rat HSC-T6 cell line was provided by Professor Lie-Ming Xu (Shanghai University of Chinese Traditional Medicine, Shanghai, China). The cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Tianjin Haoyang Biological Manufacture Co., Ltd., Tianjin, China), 100 U/ml penicillin and 100 U/ml streptomycin (Sigma-Aldrich), and incubated at 37°C in 5% CO2 air. Following 2 weeks of culture on plastic tissue-culture dishes, the cells were plated at a density of 2×104 cells/well in 96-well plates. In this experiment, these HSCs were randomly divided into three groups: Control group, PDGF-BB (R&D Systems, Inc., Minneapolis, MN, USA) group and PDGF-BB+GFK group. The concentration of PDGF-BB used for treatment was 10 ng/ml for 24 h at 37°C, and the concentrations of GFK-medicated serum (Dalian Medical University) were 2, 4, 6, 8, 10, 12, 14 and 16%. Cell viability was measured using an MTT assay (Beyotime Institute of Biotechnology, Shanghai, China) at a wavelength of 490 nm, and cellular morphology was observed using phase-contrast microscopy (Olympus IX51; Olympus Corporation).

Immunohistochemistry

The liver tissue sections were deparaffinized using xylene (Nanjing Chemical Reagent Co., Ltd.), rehydrated with graded alcohols, treated with 0.3% endogenous peroxidase blocking solution (Sigma-Aldrich) for 20 min. Following high pressure heating retrieval (125°C and 103 kPa) and non-immune goat serum (ZSGB-BIO, Beijing, China) blocking, the sections were incubated with one of the following primary antibodies purchased from BIOSS (Beijing, China): Rabbit anti-rat polyclonal anti-α-smooth muscle actin (SMA; 1:200; bs-0189R), anti-PDGF-BB (1:200; bs-1316R) and anti-PDGF receptor β (PDGFRβ; 1:150; bs-0232R). The primary antibodies were incubated overnight at 4°C. Following washing with phosphate-buffered saline (PBS), goat anti-rabbit non-biotinylated regents (ZSGB-BIO; cat. no. PV-9001) were used to react with the primary antibody for 2 h at 37°C, followed by the addition of diaminobenzidine (ZSGB-BIO) and monitoring of the staining. PBS was used as a negative control. In each section, five randomly-selected fields were examined using an Image-Pro Plus 6.0 analyzing system (Media Cybernetics, Inc., Rockville, MD, USA).

Western blotting

The manually homogenized liver tissues and cell lysates were treated with 150 μl radioimmunoprecipitation assay extraction buffer (Beyotime Institute of Biotechnology), and the supernatants were collected following centrifugation at 12,000 × g for 10 min at 4°C. The concentration of total protein was estimated using a Bradford assay, with bovine serum albumin (BSA; Sigma-Aldrich) as a standard. The protein samples (10 μg) were subjected to 12% SDS-PAGE and were then transferred onto to polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). Following blocking with 5% BSA for 3 h at room temperature, the membranes were treated with the following primary antibodies: Rabbit anti-rat polyclonal antibodies against nuclear factor of activated T cells (NFAT; 1:200; Wuhan Boster Biological Technology, Ltd., Wuhan, China) and MMP7 (1:200; Wuhan Boster Biological Technology, Ltd.) overnight at 4°C, washed with Tris-buffered saline containing 0.05% Tween 20 (TBST), and then incubated with peroxidase-conjugated secondary goat anti-rabbit antibody (1:2,000; Santa Cruz Biotechnology, Inc.; cat. no. sc-2004) for 2 h at room temperature. The membranes were washed with TBST and Prolight-horseradish peroxidase (Tiangen Biotech, Co., Ltd., Beijing, China) was used for blot detection. Rabbit anti-rat polyclonal β-actin antibodies (1:1,000; Santa Cruz Biotechnology, Inc.; cat. no. 130657) served as a loading control. Densitometric analysis was performed with LabWorks 4.6 (UVP, Inc., Upland, CA, USA).

Reverse transcription-polymerase chain reaction (RT-PCR) analysis

Total RNA was extracted from the liver tissues and cell lysates using TRIzol reagent, according to the manufacturer's protocol (Invitrogen; Thermo Fisher Scientific, Inc.). Reverse transcription with oligo (dT) primers (Tiangen Biotech, Co., Ltd.), Quant reverse transcriptase (Tiangen Biotech, Co., Ltd.) and dNTP mixture were used to synthesize complementary DNA (cDNA) from the total RNA. The primers were synthesized by Takara Biotechnology, Co., Ltd. (Dalian, China) for this purpose, and are as listed in Table II. A PCR reaction kit (Tiangen Biotech Co., Ltd.) containing 2 μl cDNA template, 12.5 μl 2X Taq PCR Master mix, 1 μl primers and 8.5 μl double distilled water to a total volume of 25 μl. Each sample had four replicates. The conditions for amplification were as follows: Wnt5a, Collagen type I, α-SMA, Calcineurin: One cycle of 94°C for 5 min, 32 cycles of 94°C for 30 sec, 64°C for 30 sec, 72°C for 30 sec, and a final extension of one cycle at 72°C for 5 min; PDGF-BB and PDGFRβ: 30 cycles of 94°C for 30 sec, 57°C for 30 sec and 72°C for 40 sec; Frizzled2: 35 cycles of 94°C for 30 sec, 57°C for 30 sec and 72°C for 35 sec; NFAT: 32 cycles of 94°C for 30 sec, 58°C for 30 sec and 72°C for 30 sec; calmodulin-dependent protein kinase II (CaMK II): 35 cycles of 94°C for 30 sec, 56°C for 30 sec and 72°C for 40 sec; Collagen type III and β-actin: 32 cycles of 94°C for 30 sec, 55°C for 30 sec and 72°C for 30 sec. The RT-PCR reaction was conducted using a T100 thermal cycler (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The PCR products were electrophoresed on 2% agarose gels (Tiangen Biotech, Co., Ltd.) and the band density was measured using LabWorks 4.6. β-actin served as a loading control.

Table II

Primer sequences and amplicon sizes.

Table II

Primer sequences and amplicon sizes.

GeneForward (5′-3′)Reverse (5′-3′)Product size (bp)
Wnt5a AGGACTTACCTCGGGACTGG TGCGACCTGCTTCATTGTT171
Frizzled2 CCTGGAGGTGCATCAATTCTAC CGCTCACCCAGAAACTTATAGC447
CaMK II TTCTACTGTTGCCTCCAT AAAGTCCATCCCTTCCAC460
Calcineurin GGACAGGGTGGTGAAAGC AGCGAGTGTTGGCAGGAG296
NFAT GCCCAGCGATGAGTATGAA ATGCACCAGCACAGAACG310
MMP-7 TGCCGGAGACTGGAAAGCTG GGTGCAAAGGCATGGCCTAG343
Collagen type I TGCCGTGACCTCAAGATGTG CACAAGCGTGCTGTAGGTGA461
Collagen type III AGATCATGTCTTGACTCAAGTC TTTACATTGCCATTGGCCTGA463
α-SMA TGTGCTGGACTCTGGAGATG GATCACCTGCCCATCAGG291
β-actin GGTATGGGTCAGAAGGACTCC TGATCTTCAGGTGCTAGGAGCC847

[i] CaMK II, calmodulin-dependent kinase II; NFAT, nuclear factor of activated T cells; MMP,-7 matrix metalloproteinase-7; α-SMA, α-smooth muscle actin.

Statistical analysis

SPSS 19.0 statistical software (IBM SPSS, Armonk, NY, USA) was used for statistical analysis. All data are expressed as the mean ± standard deviation. Comparisons were performed using one-way analysis of variance. Ordinal data were analyzed using a Kruskal-Wallis test. P<0.05 was considered to indicate a statistically significant difference.

Results

GFK treatment reduces CCl4-induced liver injury in rats

The degrees of hepatic damage in the rats were assessed by histopathological examination of the liver sections using H&E staining. As shown in Fig. 1A, compared with the control group, the livers of the CCl4 treated rats exhibited marked fatty infiltration, bridging necrosis, damage of liver lobules and wide infiltration of inflammatory cells around the central vein. The livers of the rats in the GFK (31.25 mg/kg) group showed moderate widening and a fibrotic area with bridging fibrosis, which were also observed in the GFK (3,125 mg/kg) group. However, in the livers of the GFK (312.5 mg/kg)-treated rats, only mild focal fibrotic changes of the portal area and mild bridging were observed. The histopathological scores of liver fibrosis are shown in Table III.

Table III

Histopathological semi-quantitative scores of rat liver tissues.

Table III

Histopathological semi-quantitative scores of rat liver tissues.

GroupnHepatic fibrosis score (n)
Mean score
01234
Control121200005.5b
CCl4  8  0023344.82a
CCl4+GFK (3,125)11  04430 33.17a,b
CCl4+GFK (312.5)11  07400 22.96a,b
CCl4+GFK (31.25)11  03530 31.85a,b

a P<0.05, vs. control group;

b P<0.05, vs. CCl4 group. The concentrations of 3,125, 312.5 and 31.25 are in mg/kg. CCL4, carbon tetrachloride; GFK, Gan-fu-kang.

CCl4 treatment significantly increased the serum levels of AST and ALT by four and three-fold, respectively, and markedly decreased the serum levels of TP and ALB, compared with the control group (P<0.05). The administration of GFK markedly reduced the elevated levels in serum AST (P<0.05; 3,125 and 312.5 mg/kg) and ALT (P<0.05; 312.5 mg/kg), compared with the CCl4 group. GFK treatment (312.5 mg/kg) significantly enhanced the CCl4-induced decrease in the serum level of TP (P<0.05). The serum level of ALB showed a marginal increase in the GFK-treated groups (3,125 and 312.5 mg/kg), although without statistical significance (Fig. 1B).

GFK treatment attenuates CCl4-induced collagen accumulation

Picro Sirius red staining was performed to observe collagen synthesis in the liver tissues. Collagen I was red and collagen III was green. Consistent with the H&E staining, the fibrotic changes were substantially decreased following administration of GFK at a dose of 312.5 mg/kg (Fig. 1A). Compared with the normal rats, CCl4 administration caused a significant increase in the accumulation of collagen, as assessed by the hepatic Hyp content and mRNA expression levels of collagen type I and III. In the CCl4-treated model group, the hepatic Hyp content was significantly increased, by three-fold, compared with the control group. GFK treatment notably reduced the elevated levels of Hyp, compared with the CCl4 group, when treated at GFK concentrations of 312.5 and 3,125 mg/kg (P<0.05; Fig. 2A). The results of the RT-PCR analysis showed that treatment with various doses of GFK reduced the deposition of collagen type I and III in the CCl4-induced fibrotic liver tissues (Fig. 2B).

GFK treatment suppresses α-SMA activation, and the expression levels of PDGF-BB and PDGFRβ

α-SMA is a marker used to evaluate the degree of HSC activation (23). In addition, PDGF is one of the most potent mitogens for HSCs. Accordingly, PDGFRβ is expressed in HSCs when transformed, and may be a marker of HSC activation. In the present study, RT-PCR analysis revealed that the upregulation in the mRNA levels of α-SMA, PDGF-BB and PDGFRβ induced by CCl4 were significantly attenuated by GFK (Fig. 3A). Image analysis of the immunohistochemical staining of α-SMA, PDGF-BB and PDGFRβ showed that, compared with the control group, CCl4 treatment significantly increased the accumulation of activated HSCs (P<0.01; Fig. 3B). Treatment with the different doses of GFK significantly decreased HSC activation in the liver (P<0.01), with the medium dose (312.5 mg/kg) having the most pronounced effect on HSC activation (Fig. 3B).

GFK treatment inhibits activation of the Wnt/Ca2+ signaling pathway in vivo

To elucidate the possible molecular pathway by which GFK suppressed liver fibrosis, the present study examined the mRNA and protein expression levels of several associated genes in the Wnt/Ca2+ signaling pathway. The mRNA expression levels of Wnt5a, Frizzled2, CaMK II, calcineurin, NFAT and MMP-7 were increased in the CCl4-treated livers, compared with the control group. Following administration of GFK, the mRNA expression levels decreased at GFK dose of 312.5 mg/kg, compared with the CCl4 group (Fig. 4A). Western blot analysis indicated a significant reduction in the protein levels of NFAT and MMP-7 (P<0.01; Fig. 4B). This suggested that GFK inhibits the Wnt/Ca2+ signaling pathway via protein synthesis levels rather than by repressing mRNA expression.

GFK treatment attenuates the proliferation and activation of HSC-T6 cells

To evaluate the effects of GFK on HSCs in the liver, HSC-T6 cells were treated with increasing concentrations of GFK. The MTT assay showed that GFK treatment caused a dose-dependent reduction in the number of HSC-T6 cells (Fig. 5A). The results revealed that GFK inhibited the viability, which was stimulated by PDGF-BB. The present study then calculated the half maximal inhibitory concentration (IC50), which was used in the subsequent experiment. As shown in Fig. 5B, the HSCs cultured with PDGF-BB for 24 h exhibited an intermediate stage of the activation process. The HSCs exposed to GFK at the IC50 maintained a quiescent morphology. In addition, GFK treatment at the IC50 markedly decreased the expression levels of PDGFRβ, α-SMA, and collagen type I and III (Fig. 5C).

GFK treatment inhibits the Wnt/Ca2+ signaling pathway in HSC-T6 cells

The levels of principal elements in the Wnt/Ca2+ signaling pathway, including Wnt5a, Frizzled2, CaMK II, calcineurin, NFAT and MMP-7, were detected in HSC-T6 cells using RT-PCR and Western blot analyses. The results showed that the expression levels of these components were all attenuated in the presence of GFK at the IC50 (Fig. 6).

Discussion

At present, the major obstacles in the treatment of liver fibrosis are the unsatisfactory effects and various side effects due to immune suppression and cytotoxicity (24). Medicinally, herbal drugs often provide alternative treatment options for liver diseases. GFK is a multi-ingredient herbal drug, which contains 11 crude plant ingredients. These 11 plants offer certain synergistic effects in hepatic injury therapy. One of the important components of GFK is Astragalus membranaceus, which has been found to significantly delay the formation of liver fibrosis induced by CCl4 in vivo (25). The other major component in GFK, Salvia miltiorrhiza, can induce HSC apoptosis or inhibit the proliferation of HSCs (26). The remaining components of GFK, including Atractylodes macrocephala, Rehmannia glutinosa and licorice root, have been confirmed to have hepatoprotective effects (2729). In the present study, the results showed that GFK suppresses the progress of CCl4-induced liver fibrosis in rats. The data also indicated that GFK inhibited the proliferation of HSCs.

In the present study, as expected, histopathological examination (H&E staining) revealed that an 8-week period of subcutaneous injection of CCl4 (0.5 mg/kg; twice per week) markedly induced hepatofibrotic changes, whereas GFK administration substantially ameliorate these alternations. These findings were also confirmed by the detection of the biochemical indicators, ALT, AST, TP and ALB. The progressive accumulation of ECM results in liver fibrosis. Collagen, particularly collagen type I and type III is the predominant component of the ECM, and Hyp is a degradation product of collagen, which has been used as an indicator for evaluating collagen deposition (30). The results of the present study showed that CCl4 notably induced the accumulation of collagen, as determined by Hyp concentrations and Picric Sirius red staining, and that collagen deposition was markedly lowered by GFK treatment. It was also found that GFK administration significantly downregulated the expression levels of collagen type I and type III in vivo and in vitro, suggesting that GFK may enhance collagenolysis in the fibrotic liver.

It is generally accepted that HSCs are pivotal in the development of liver fibrosis, and that α-SMA is a key marker of HSC activation. In the present study, GFK was found to decrease HSC viability in a dose-dependent manner. Additionally, the data confirmed that GFK treatment (312.5 mg/kg) considerably inhibited the activation of HSCs, as observed in the immunohistochemical staining of α-SMA. The suppression of the gene expression levels of α-SMA in liver fibrosis and in the PDGF-BB-treated HSC-T6 cells were also more prominent in the GFK administration group. Consistent with the changes in levels of α-SMA, the expression levels of the profibrogenic cytokines, PDGF-BB and PDGFRβ, were marked in the fibrotic liver tissues, whereas they were attenuated following GFK treatment. In addition, GFK decreased the protein and mRNA levels of PDGF-BB and PDGFRβ in vivo and in vitro, demonstrating that the antifibrotic effects of GFK exerted its inhibitory effects on HSC activation via modulation of the profibrogenic cytokines, PDGF-BB and PDGFRβ.

The activation of Wnt signaling has been implicated in the conversion of numerous fibrotic diseases, including lung, heart, kidney and liver fibrosis (3134). It has been demonstrated that the canonical and non-canonical Wnt signaling pathways are involved in the pathogenesis of liver fibrosis (35). The non-canonical Wnt/Ca2+ pathway includes the binding of Wnt5a to its receptor, Frizzled2, which then leads to elevated intracellular Ca2+ concentrations via a G-protein-dependent mechanism (36). Elevated Ca2+ can activate Ca2+/CaMK II via calcineurin/NFAT pathways (3739). The transcription factor, NFAT, translocates from the cytosol to the nucleus, thereby activating downstream target gene transcription (40). In the present study, the expression levels of components of the Wnt/Ca2+ signaling pathway, including Wnt5a, Frizzled2, CaMK II, calcineurin, NFAT and MMP-7, were markedly upregulated in the rats with hepatic fibrosis and the PDGF-BB-treated HSC-T6 cells. However, they were markedly decreased by GFK treatment. Furthermore, the effects on the protein levels of NFAT and MMP-7 were in accordance with the alterations in their mRNA expression levels. Taken together, these results suggested that GFK ameliorated liver fibrosis and activated HSCs by regulating the Wnt/Ca2+ signaling pathway.

Acknowledgments

This study was financially supported by the Natural Science Foundation of Liaoning Province, China (grant no. 201102055).

References

1 

Friedman SL, Maher JJ and Bissell DM: Mechanisms and therapy of hepatic fibrosis: Report of the AASLD single topic basic research conference. Hepatology. 32:1403–1408. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Pellicoro A, Ramachandran P, Iredale JP and Fallowfield JA: Liver fibrosis and repair: Immune regulation of wound healing in a solid organ. Nat Rev Immunol. 14:181–194. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Puche JE, Saiman Y and Friedman SL: Hepatic stellate cells and liver fibrosis. Compr Physiol. 3:1473–1492. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Duval F, Moreno-Cuevas JE, González-Garza MT, Rodríguez-Montalvo C and Cruz-Vega DE: Liver fibrosis and protection mechanisms action of medicinal plants targeting apoptosis of hepatocytes and hepatic stellate cells. Adv Pharmacol Sci. 2014:3732952014.PubMed/NCBI

5 

Breitkopf K, Roeyen CV, Sawitza I, Wickert L, Floege J and Gressner AM: Expression patterns of PDGF-A, -B, -C and -D and the PDGF-receptors alpha and beta in activated rat hepatic stellate cells (HSC). Cytokine. 31:349–357. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Fang L, Zhan S, Huang C, Cheng X, Lv X, Si H and Li J: TRPM7 channel regulates PDGF-BB-induced proliferation of hepatic stellate cells via PI3 K and ERK pathways. Toxicol Appl Pharmacol. 272:713–725. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Shah R, Reyes-Gordillo K, Arellanes-Robledo J, Lechuga CG, Hernández-Nazara Z, Cotty A, Rojkind M and Lakshman MR: TGF-β 1 up-regulates the expression of PDGF-β receptor mRNA and induces a delayed PI3K-, AKT- and p70 S6K-dependent proliferative response in activated hepatic stellate cells. Alcohol Clin Exp Res. 37:1838–1848. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Miao CG, Yang YY, He X, Huang C, Huang Y, Zhang L, Lv XW, Jin Y and Li J: Wnt signaling in liver fibrosis: Progress, challenges and potential directions. Biochimie. 95:2326–2335. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Al-Harthi L: Wnt/β-catenin and its diverse physiological cell signaling pathways in neurodegenerative and neuropsychiatric disorders. J Neuroimmune Pharmacol. 7:725–730. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Holland JD, Klaus A, Garratt AN and Birchmeier W: Wnt signaling in stem and cancer stem cells. Curr Opin Cell Biol. 25:254–264. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Ge WS, Wang YJ, Wu JX, Fan JG, Chen YW and Zhu L: β-catenin is overexpressed in hepatic fibrosis and blockage of Wnt/β-catenin signaling inhibits hepatic stellate cell activation. Mol Med Rep. 9:2145–2151. 2014.PubMed/NCBI

12 

Rashid ST, Humphries JD, Byron A, Dhar A, Askari JA, Selley JN, Knight D, Goldin RD, Thursz M and Humphries MJ: Proteomic analysis of extracellular matrix from the hepatic stellate cell line LX-2 identifies CYR61 and Wnt-5a as novel constituents of fibrotic liver. J Proteome Res. 11:4052–4064. 2012. View Article : Google Scholar : PubMed/NCBI

13 

MadanKumar P, NaveenKumar P, Manikandan S, Devaraj H and NiranjaliDevaraj S: Morin ameliorates chemically induced liver fibrosis in vivo and inhibits stellate cell proliferation in vitro by suppressing Wnt/β-catenin signaling. Toxicol Appl Pharmacol. 277:210–220. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Shin HW, Park SY, Lee KB, Shin E, Nam SW, Lee JY and Jang JJ: Transcriptional profiling and Wnt signaling activation in proliferation of human hepatic stellate cells induced by PDGF-BB. Korean J Hepatol. 15:486–495. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Saneyoshi T, Kume S, Amasaki Y and Mikoshiba K: The Wnt/calcium pathway activates NF-AT and promotes ventral cell fate in Xenopus embryos. Nature. 417:295–299. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Wei F, Lang Y, Gong D and Fan Y: Effect of Dahuang zhechong formula on liver fibrosis in patients with chronic hepatitis B: A meta-analysis. Complement Ther Med. 23:129–138. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Lin HJ, Chen JY, Lin CF, Kao ST, Cheng JC, Chen HL and Chen CM: Hepatoprotective effects of Yi Guan Jian, an herbal medicine, in rats with dimethylnitrosamine-induced liver fibrosis. J Ethnopharmacol. 134:953–960. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Xu TT, Jiang MN, Li C, Che Y and Jia YJ: Effect of Chinese traditional compound, Gan-fu-kang, on CCl(4)-induced liver fibrosis in rats and its probable molecular mechanisms. Hepatol Res. 37:221–229. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Lou JL, Jiang MN, Li C, Zhou Q, He X, Lei HY, Li J and Jia YJ: Herb medicine Gan-fu-kang attenuates liver injury in a rat fibrotic model. J Ethnopharmacol. 128:131–138. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Zhang C, Wang Y, Chen H, Yang G, Wang S, Jiang M, Cong L, Yuan L, Li H and Jia Y: Protective effect of the herbal medicine Gan-fu-kang against carbon tetrachloride-induced liver fibrosis in rats. Mol Med Rep. 8:954–962. 2013.PubMed/NCBI

21 

National Institutes of Health: Guide for the Care and Use of Laboratory Animals. 6th edition. National Academies Press; Washington, DC: 1985

22 

Scheuer PJ: Classification of chronic viral hepatitis: A need for reassessment. J Hepatol. 13:372–374. 1991. View Article : Google Scholar : PubMed/NCBI

23 

Zakaria S, Youssef M, Moussa M, Akl M, El-Ahwany E, El-Raziky M, Mostafa O, Helmy AH and El-Hindawi A: Value of α-smooth muscle actin and glial fibrillary acidic protein in predicting early hepatic fibrosis in chronic hepatitis C virus infection. Arch Med Sci. 6:356–365. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Schiavon F: Transient joint effusion: A forgotten side effect of high dose corticosteroid treatment. Ann Rheum Dis. 62:491–492. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Sun WY, Wang L, Liu H, Li X and Wei W: A standardized extract from Paeonia lactiflora and Astragalus membranaceus attenuates liver fibrosis induced by porcine serum in rats. Int J Mol Med. 29:491–498. 2012.

26 

Lee HS, Son WC, Ryu JE, Koo BA and Kim YS: Standardized Salvia miltiorrhiza extract suppresses hepatic stellate cell activation and attenuates steatohepatitis induced by a methionine-choline deficient diet in mice. Molecules. 19:8189–8211. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Jin C, Zhang PJ, Bao CQ, Gu YL, Xu BH, Li CW, Li JP, Bo P and Liu XN: Protective effects of Atractylodes macrocephala polysaccharide on liver ischemia-reperfusion injury and its possible mechanism in rats. Am J Chin Med. 39:489–502. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Wu PS, Wu SJ, Tsai YH, Lin YH and Chao JC: Hot water extracted Lycium barbarum and Rehmannia glutinosa inhibit liver inflammation and fibrosis in rats. Am J Chin Med. 39:1173–1191. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Hajiaghamohammadi AA, Ziaee A and Samimi R: The efficacy of licorice root extract in decreasing transaminase activities in non-alcoholic fatty liver disease: A randomized controlled clinical trial. Phytother Res. 26:1381–1384. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Hofman K, Hall B, Cleaver H and Marshall S: High-throughput quantification of hydroxyproline for determination of collagen. Anal Biochem. 417:289–291. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Song P, Zheng JX, Xu J, Liu JZ, Wu LY and Liu C: β-catenin induces A549 alveolar epithelial cell mesenchymal transition during pulmonary fibrosis. Mol Med Rep. 11:2703–2710. 2015.

32 

Ye B, Ge Y, Perens G, Hong L, Xu H, Fishbein MC and Li F: Canonical Wnt/β-catenin signaling in epicardial fibrosis of failed pediatric heart allografts with diastolic dysfunction. Cardiovasc Pathol. 22:54–57. 2013. View Article : Google Scholar

33 

Li X, Yamagata K, Nishita M, Endo M, Arfian N, Rikitake Y, Emoto N, Hirata K, Tanaka Y and Minami Y: Activation of Wnt5a-Ror2 signaling associated with epithelial-to-mesenchymal transition of tubular epithelial cells during renal fibrosis. Genes Cells. 18:608–619. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Li W, Zhu C, Li Y, Wu Q and Gao R: Mest attenuates CCl4-induced liver fibrosis in rats by inhibiting the Wnt/β-catenin signaling pathway. Gut Liver. 8:282–291. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Jiang F, Parsons CJ and Stefanovic B: Gene expression profile of quiescent and activated rat hepatic stellate-cells implicates Wnt signaling pathway in activation. J Hepatol. 45:401–409. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Sheldahl LC, Park M, Malbon CC and Moon RT: Protein kinase C is differentially stimulated by Wnt and Frizzled homologs in a G-protein-dependent manner. Curr Biol. 9:695–698. 1999. View Article : Google Scholar : PubMed/NCBI

37 

De A: Wnt/Ca2+ signaling pathway: A brief overview. Acta Biochim Biophys Sin (Shanghai). 43:745–756. 2011. View Article : Google Scholar

38 

Buchholz M, Schatz A, Wagner M, Michl P, Linhart T, Adler G, Gress TM and Ellenrieder V: Overexpression of c-myc in pancreatic cancer caused by ectopic activation of NFATc1 and the Ca2+/calcineurin signaling pathway. EMBO J. 25:3714–3724. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Kohn AD and Moon RT: Wnt and calcium signaling: Beta-catenin-independent pathways. Cell Calcium. 38:439–446. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Kim J, Kim DW, Chang W, Choe J, Kim J, Park CS, Song K and Lee I: Wnt5a is secreted by follicular dendritic cells to protect germinal center B cells via Wnt/Ca2+/NFAT/NF-κB-B cell lymphoma 6 signaling. J Immunol. 188:182–189. 2012. View Article : Google Scholar

Related Articles

Journal Cover

June-2016
Volume 13 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jia Y, Yuan L, Xu T, Li H, Yang G, Jiang M, Zhang C and Li C: Herbal medicine Gan‑fu‑kang downregulates Wnt/Ca2+ signaling to attenuate liver fibrogenesis in vitro and in vivo. Mol Med Rep 13: 4705-4714, 2016
APA
Jia, Y., Yuan, L., Xu, T., Li, H., Yang, G., Jiang, M. ... Li, C. (2016). Herbal medicine Gan‑fu‑kang downregulates Wnt/Ca2+ signaling to attenuate liver fibrogenesis in vitro and in vivo. Molecular Medicine Reports, 13, 4705-4714. https://doi.org/10.3892/mmr.2016.5148
MLA
Jia, Y., Yuan, L., Xu, T., Li, H., Yang, G., Jiang, M., Zhang, C., Li, C."Herbal medicine Gan‑fu‑kang downregulates Wnt/Ca2+ signaling to attenuate liver fibrogenesis in vitro and in vivo". Molecular Medicine Reports 13.6 (2016): 4705-4714.
Chicago
Jia, Y., Yuan, L., Xu, T., Li, H., Yang, G., Jiang, M., Zhang, C., Li, C."Herbal medicine Gan‑fu‑kang downregulates Wnt/Ca2+ signaling to attenuate liver fibrogenesis in vitro and in vivo". Molecular Medicine Reports 13, no. 6 (2016): 4705-4714. https://doi.org/10.3892/mmr.2016.5148